Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 173(1): 140-152.e15, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29570993

RESUMEN

Hunger and pain are two competing signals that individuals must resolve to ensure survival. However, the neural processes that prioritize conflicting survival needs are poorly understood. We discovered that hunger attenuates behavioral responses and affective properties of inflammatory pain without altering acute nociceptive responses. This effect is centrally controlled, as activity in hunger-sensitive agouti-related protein (AgRP)-expressing neurons abrogates inflammatory pain. Systematic analysis of AgRP projection subpopulations revealed that the neural processing of hunger and inflammatory pain converge in the hindbrain parabrachial nucleus (PBN). Strikingly, activity in AgRP → PBN neurons blocked the behavioral response to inflammatory pain as effectively as hunger or analgesics. The anti-nociceptive effect of hunger is mediated by neuropeptide Y (NPY) signaling in the PBN. By investigating the intersection between hunger and pain, we have identified a neural circuit that mediates competing survival needs and uncovered NPY Y1 receptor signaling in the PBN as a target for pain suppression.


Asunto(s)
Neuronas/metabolismo , Dolor/patología , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Analgésicos Opioides/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Conducta Animal/efectos de los fármacos , Dieta , Conducta Alimentaria/efectos de los fármacos , Formaldehído/toxicidad , Glutamato Descarboxilasa/metabolismo , Locomoción/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Morfina/farmacología , Neuronas/efectos de los fármacos , Dolor/etiología , Dolor/metabolismo , Núcleos Parabraquiales/efectos de los fármacos , Núcleos Parabraquiales/metabolismo , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal
2.
Cell ; 155(6): 1337-50, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24315102

RESUMEN

Neural circuits for essential natural behaviors are shaped by selective pressure to coordinate reliable execution of flexible goal-directed actions. However, the structural and functional organization of survival-oriented circuits is poorly understood due to exceptionally complex neuroanatomy. This is exemplified by AGRP neurons, which are a molecularly defined population that is sufficient to rapidly coordinate voracious food seeking and consumption behaviors. Here, we use cell-type-specific techniques for neural circuit manipulation and projection-specific anatomical analysis to examine the organization of this critical homeostatic circuit that regulates feeding. We show that AGRP neuronal circuits use a segregated, parallel, and redundant output configuration. AGRP neuron axon projections that target different brain regions originate from distinct subpopulations, several of which are sufficient to independently evoke feeding. The concerted anatomical and functional analysis of AGRP neuron projection populations reveals a constellation of core forebrain nodes, which are part of an extended circuit that mediates feeding behavior.


Asunto(s)
Encéfalo/fisiología , Conducta Alimentaria , Homeostasis , Vías Nerviosas , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Ghrelina/metabolismo , Ratones
3.
Nature ; 612(7941): 739-747, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36517598

RESUMEN

Exercise exerts a wide range of beneficial effects for healthy physiology1. However, the mechanisms regulating an individual's motivation to engage in physical activity remain incompletely understood. An important factor stimulating the engagement in both competitive and recreational exercise is the motivating pleasure derived from prolonged physical activity, which is triggered by exercise-induced neurochemical changes in the brain. Here, we report on the discovery of a gut-brain connection in mice that enhances exercise performance by augmenting dopamine signalling during physical activity. We find that microbiome-dependent production of endocannabinoid metabolites in the gut stimulates the activity of TRPV1-expressing sensory neurons and thereby elevates dopamine levels in the ventral striatum during exercise. Stimulation of this pathway improves running performance, whereas microbiome depletion, peripheral endocannabinoid receptor inhibition, ablation of spinal afferent neurons or dopamine blockade abrogate exercise capacity. These findings indicate that the rewarding properties of exercise are influenced by gut-derived interoceptive circuits and provide a microbiome-dependent explanation for interindividual variability in exercise performance. Our study also suggests that interoceptomimetic molecules that stimulate the transmission of gut-derived signals to the brain may enhance the motivation for exercise.


Asunto(s)
Eje Cerebro-Intestino , Dopamina , Ejercicio Físico , Microbioma Gastrointestinal , Motivación , Carrera , Animales , Ratones , Encéfalo/citología , Encéfalo/metabolismo , Dopamina/metabolismo , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/metabolismo , Células Receptoras Sensoriales/metabolismo , Eje Cerebro-Intestino/fisiología , Microbioma Gastrointestinal/fisiología , Ejercicio Físico/fisiología , Ejercicio Físico/psicología , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/psicología , Modelos Animales , Humanos , Estriado Ventral/citología , Estriado Ventral/metabolismo , Carrera/fisiología , Carrera/psicología , Recompensa , Individualidad
4.
Nature ; 600(7888): 269-273, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34789878

RESUMEN

The brain is the seat of body weight homeostasis. However, our inability to control the increasing prevalence of obesity highlights a need to look beyond canonical feeding pathways to broaden our understanding of body weight control1-3. Here we used a reverse-translational approach to identify and anatomically, molecularly and functionally characterize a neural ensemble that promotes satiation. Unbiased, task-based functional magnetic resonance imaging revealed marked differences in cerebellar responses to food in people with a genetic disorder characterized by insatiable appetite. Transcriptomic analyses in mice revealed molecularly and topographically -distinct neurons in the anterior deep cerebellar nuclei (aDCN) that are activated by feeding or nutrient infusion in the gut. Selective activation of aDCN neurons substantially decreased food intake by reducing meal size without compensatory changes to metabolic rate. We found that aDCN activity terminates food intake by increasing striatal dopamine levels and attenuating the phasic dopamine response to subsequent food consumption. Our study defines a conserved satiation centre that may represent a novel therapeutic target for the management of excessive eating, and underscores the utility of a 'bedside-to-bench' approach for the identification of neural circuits that influence behaviour.


Asunto(s)
Mantenimiento del Peso Corporal/genética , Mantenimiento del Peso Corporal/fisiología , Cerebelo/fisiología , Alimentos , Biosíntesis de Proteínas , Genética Inversa , Respuesta de Saciedad/fisiología , Adulto , Animales , Regulación del Apetito/genética , Regulación del Apetito/fisiología , Núcleos Cerebelosos/citología , Núcleos Cerebelosos/fisiología , Cerebelo/citología , Señales (Psicología) , Dopamina/metabolismo , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Femenino , Homeostasis , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Neostriado/metabolismo , Neuronas/fisiología , Obesidad/genética , Filosofía , Adulto Joven
5.
Cell ; 139(1): 161-74, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19804761

RESUMEN

GABAergic interneurons are key elements in neural coding, but the mechanisms that assemble inhibitory circuits remain unclear. In the spinal cord, the transfer of sensory signals to motor neurons is filtered by GABAergic interneurons that act presynaptically to inhibit sensory transmitter release and postsynaptically to inhibit motor neuron excitability. We show here that the connectivity and synaptic differentiation of GABAergic interneurons that mediate presynaptic inhibition is directed by their sensory targets. In the absence of sensory terminals these GABAergic neurons shun other available targets, fail to undergo presynaptic differentiation, and withdraw axons from the ventral spinal cord. A sensory-specific source of brain derived neurotrophic factor induces synaptic expression of the GABA synthetic enzyme GAD65--a defining biochemical feature of this set of interneurons. The organization of a GABAergic circuit that mediates presynaptic inhibition in the mammalian CNS is therefore controlled by a stringent program of sensory recognition and signaling.


Asunto(s)
Interneuronas/fisiología , Médula Espinal/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Glutamato Descarboxilasa , Ratones , Neuronas Motoras/fisiología , Terminales Presinápticos , Propiocepción , Células Receptoras Sensoriales/fisiología , Médula Espinal/citología
6.
Nature ; 521(7551): 180-185, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25915020

RESUMEN

Homeostasis is a biological principle for regulation of essential physiological parameters within a set range. Behavioural responses due to deviation from homeostasis are critical for survival, but motivational processes engaged by physiological need states are incompletely understood. We examined motivational characteristics of two separate neuron populations that regulate energy and fluid homeostasis by using cell-type-specific activity manipulations in mice. We found that starvation-sensitive AGRP neurons exhibit properties consistent with a negative-valence teaching signal. Mice avoided activation of AGRP neurons, indicating that AGRP neuron activity has negative valence. AGRP neuron inhibition conditioned preference for flavours and places. Correspondingly, deep-brain calcium imaging revealed that AGRP neuron activity rapidly reduced in response to food-related cues. Complementary experiments activating thirst-promoting neurons also conditioned avoidance. Therefore, these need-sensing neurons condition preference for environmental cues associated with nutrient or water ingestion, which is learned through reduction of negative-valence signals during restoration of homeostasis.


Asunto(s)
Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Hambre/fisiología , Neuronas/metabolismo , Sed/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Señales (Psicología) , Deshidratación , Alimentos , Preferencias Alimentarias , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Modelos Animales , Inanición
8.
Nature ; 488(7410): 172-7, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22801496

RESUMEN

Hunger is a complex behavioural state that elicits intense food seeking and consumption. These behaviours are rapidly recapitulated by activation of starvation-sensitive AGRP neurons, which present an entry point for reverse-engineering neural circuits for hunger. Here we mapped synaptic interactions of AGRP neurons with multiple cell populations in mice and probed the contribution of these distinct circuits to feeding behaviour using optogenetic and pharmacogenetic techniques. An inhibitory circuit with paraventricular hypothalamus (PVH) neurons substantially accounted for acute AGRP neuron-evoked eating, whereas two other prominent circuits were insufficient. Within the PVH, we found that AGRP neurons target and inhibit oxytocin neurons, a small population that is selectively lost in Prader-Willi syndrome, a condition involving insatiable hunger. By developing strategies for evaluating molecularly defined circuits, we show that AGRP neuron suppression of oxytocin neurons is critical for evoked feeding. These experiments reveal a new neural circuit that regulates hunger state and pathways associated with overeating disorders.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Hambre/fisiología , Vías Nerviosas/fisiología , Neuronas/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Axones/metabolismo , Femenino , Privación de Alimentos , Masculino , Ratones , Modelos Neurológicos , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proopiomelanocortina/metabolismo , Inanición , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/metabolismo
9.
bioRxiv ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38464066

RESUMEN

Long-term sustained pain in the absence of acute physical injury is a prominent feature of chronic pain conditions. While neurons responding to noxious stimuli have been identified, understanding the signals that persist without ongoing painful stimuli remains a challenge. Using an ethological approach based on the prioritization of adaptive survival behaviors, we determined that neuropeptide Y (NPY) signaling from multiple sources converges on parabrachial neurons expressing the NPY Y1 receptor to reduce sustained pain responses. Neural activity recordings and computational modeling demonstrate that activity in Y1R parabrachial neurons is elevated following injury, predicts functional coping behavior, and is inhibited by competing survival needs. Taken together, our findings suggest that parabrachial Y1 receptor-expressing neurons are a critical hub for endogenous analgesic pathways that suppress sustained pain states.

10.
Artículo en Inglés | MEDLINE | ID: mdl-39010249

RESUMEN

In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.

11.
Cell Rep ; 42(11): 113338, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37910501

RESUMEN

Caloric restriction has anti-inflammatory effects. However, the coordinated physiological actions that lead to reduced inflammation in a state of caloric deficit (hunger) are largely unknown. Using a mouse model of injury-induced peripheral inflammation, we find that food deprivation reduces edema, temperature, and cytokine responses that occur after injury. The magnitude of the anti-inflammatory effect that occurs during hunger is more robust than that of non-steroidal anti-inflammatory drugs. The effects of hunger are recapitulated centrally by activity in nutrient-sensing hypothalamic agouti-related protein (AgRP)-expressing neurons. We find that AgRP neurons projecting to the paraventricular nucleus of the hypothalamus rapidly and robustly reduce inflammation and mediate the majority of hunger's anti-inflammatory effects. Intact vagal efferent signaling is required for the anti-inflammatory action of hunger, revealing a brain-to-periphery pathway for this reduction in inflammation. Taken together, these data begin to unravel a potent anti-inflammatory pathway engaged by hypothalamic AgRP neurons to reduce inflammation.


Asunto(s)
Hambre , Hipotálamo , Humanos , Hambre/fisiología , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Inflamación/metabolismo
12.
Neuroscience ; 483: 40-51, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34923039

RESUMEN

The mesolimbic dopamine (DA) system reinforces behaviors that are critical for survival. However, drug dependence can occur when drugs of abuse, such as nicotine, highjack this reinforcement system. Pharmacologically targeting the DA system to selectively block drug reinforcement requires a detailed understanding of the neural circuits and molecular pathways that lead to the reward-based activation of mesolimbic circuits. Varenicline is an approved smoking cessation drug that has been shown to block nicotine-evoked DA increases in the nucleus accumbens (NAc) through action on nicotinic acetylcholine receptors. Because these receptors have been implicated in the reinforcement of other addictive substances, we explored the possibility that varenicline could broadly affect reward processing. We used in vivo fiber photometry to monitor midbrain DA neuron activity and striatal DA levels following either natural or drug rewards in mice treated with varenicline. We demonstrate that varenicline pretreatment enhances the suppression of nicotine-evoked DA release by attenuating DA neuron activity in the VTA. Varenicline's ability to attenuate DA release is highly specific to nicotine, and varenicline slightly elevates DA release when co-administered with morphine or ethanol. Furthermore, varenicline has no effect on DA release in response to naturally rewarding behavior such as food intake or exercise. These results demonstrate the exquisite specificity with which varenicline blocks nicotine reward and highlight the complexity with which different rewards activate the mesolimbic DA system.


Asunto(s)
Agonistas Nicotínicos , Preparaciones Farmacéuticas , Animales , Dopamina/metabolismo , Ratones , Agonistas Nicotínicos/farmacología , Núcleo Accumbens/metabolismo , Preparaciones Farmacéuticas/metabolismo , Recompensa , Vareniclina/metabolismo , Vareniclina/farmacología , Vareniclina/uso terapéutico
13.
Cell Metab ; 33(3): 676-687.e5, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33450178

RESUMEN

Food intake is tightly regulated by complex and coordinated gut-brain interactions. Nutrients rapidly modulate activity in key populations of hypothalamic neurons that regulate food intake, including hunger-sensitive agouti-related protein (AgRP)-expressing neurons. Because individual macronutrients engage specific receptors in the gut to communicate with the brain, we reasoned that macronutrients may utilize different pathways to reduce activity in AgRP neurons. Here, we revealed that AgRP neuron activity in hungry mice is inhibited by site-specific intestinal detection of different macronutrients. We showed that vagal gut-brain signaling is required for AgRP neuron inhibition by fat. In contrast, spinal gut-brain signaling relays the presence of intestinal glucose. Further, we identified glucose sensors in the intestine and hepatic portal vein that mediate glucose-dependent AgRP neuron inhibition. Therefore, distinct pathways are activated by individual macronutrients to inhibit AgRP neuron activity.


Asunto(s)
Intestinos/fisiología , Neuronas/metabolismo , Nutrientes/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Grasas de la Dieta/metabolismo , Grasas de la Dieta/farmacología , Glucosa/metabolismo , Glucosa/farmacología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nutrientes/farmacología , Transducción de Señal/efectos de los fármacos , Transportador 1 de Sodio-Glucosa/metabolismo , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología
14.
Mol Metab ; 54: 101352, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34626854

RESUMEN

OBJECTIVE: Long-acting glucagon-like peptide-1 receptor agonists (GLP-1RAs), like liraglutide and semaglutide, are viable treatments for diabetes and obesity. Liraglutide directly activates hypothalamic proopiomelanocortin (POMC) neurons while indirectly inhibiting Neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons ex vivo. While temporal control of GLP-1R agonist concentration as well as accessibility to tissues/cells can be achieved with relative ease ex vivo, in vivo this is dependent upon the pharmacokinetics of these agonists and relative penetration into structures of interest. Thus, whether liraglutide or semaglutide modifies the activity of POMC and NPY/AgRP neurons in vivo as well as mechanisms required for any changes in cellular activity remains undefined. METHODS: In order to resolve this issue, we utilized neuron-specific transgenic mouse models to examine changes in the activity of POMC and NPY/AgRP neurons after injection of either liraglutide or semaglutide (intraperitoneal - I.P. and subcutaneous - S·C.). POMC and NPY/AgRP neurons were targeted for patch-clamp electrophysiology as well as in vivo fiber photometry. RESULTS: We found that liraglutide and semaglutide directly activate and increase excitatory tone to POMC neurons in a time-dependent manner. This increased activity of POMC neurons required GLP-1Rs in POMC neurons as well as a downstream mixed cation channel comprised of TRPC5 subunits. We also observed an indirect upregulation of excitatory input to POMC neurons originating from glutamatergic cells that also required TRPC5 subunits. Conversely, GLP-1Ra's decreased excitatory input to and indirectly inhibited NPY/AgRP neurons through activation of K-ATP and TRPC5 channels in GABAergic neurons. Notably, the temporal activation of POMC and inhibition of NPY/AgRP neuronal activity after liraglutide or semaglutide was injected [either intraperitoneal (I.P.) or subcutaneous (S·C.)] was dependent upon the nutritional state of the animals (fed vs food-deprived). CONCLUSIONS: Our results support a mechanism of liraglutide and semaglutide in vivo to activate POMC while inhibiting NPY/AgRP neurons, which depends upon metabolic state and mirrors the pharmacokinetic profile of these compounds in vivo.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Péptidos Similares al Glucagón/farmacología , Liraglutida/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Animales , Receptor del Péptido 1 Similar al Glucagón/agonistas , Péptidos Similares al Glucagón/administración & dosificación , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Liraglutida/administración & dosificación , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/metabolismo , Factores de Tiempo
15.
Nat Neurosci ; 24(12): 1699-1710, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34795450

RESUMEN

The striatum comprises multiple subdivisions and neural circuits that differentially control motor output. The islands of Calleja (IC) contain clusters of densely packed granule cells situated in the ventral striatum, predominantly in the olfactory tubercle (OT). Characterized by expression of the D3 dopamine receptor, the IC are evolutionally conserved, but have undefined functions. Here, we show that optogenetic activation of OT D3 neurons robustly initiates self-grooming in mice while suppressing other ongoing behaviors. Conversely, optogenetic inhibition of these neurons halts ongoing grooming, and genetic ablation reduces spontaneous grooming. Furthermore, OT D3 neurons show increased activity before and during grooming and influence local striatal output via synaptic connections with neighboring OT neurons (primarily spiny projection neurons), whose firing rates display grooming-related modulation. Our study uncovers a new role of the ventral striatum's IC in regulating motor output and has important implications for the neural control of grooming.


Asunto(s)
Islotes Olfatorios , Estriado Ventral , Animales , Cuerpo Estriado/metabolismo , Aseo Animal , Ratones , Neuronas/fisiología , Tubérculo Olfatorio
16.
Neuroscience ; 450: 126-134, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32540365

RESUMEN

Unpleasant somatosensory stimuli such as pain and itch can interrupt normal behavior. But survival can depend on resuming normal behavior before these challenges are fully resolved. The neural mechanisms that prioritize behavior when individuals are challenged with unpleasant somatosensory sensations, however, are not fully understood. Recently, we identified a neural circuit activated by hunger that can inhibit pain, prioritizing food seeking over tending to an injury. Here, we examine the ability of hunger, and neurons activated by hunger, to inhibit behavioral responses to another unpleasant somatosensory sensation - itch. We demonstrate that food deprivation inhibits scratching induced by three different pruritogenic stimuli: histamine, serotonin, and chloroquine. The inhibition of scratching correlates with the level of food deprivation, suggesting a cross-competition of alarm systems in the brain whereby more energy need more efficiently inhibits competing drives. Finally, we show that activity in hunger-sensitive, hypothalamic agouti-related protein (AgRP)-expressing neurons is sufficient to inhibit itch. Taken together, we showed that hunger or AgRP neuron activity inhibits itch, demonstrating that organisms have neural systems to filter and process ascending spinal signals activated by unpleasant somatosensory stimuli to prioritize salient needs.


Asunto(s)
Hambre , Neuronas , Proteína Relacionada con Agouti/metabolismo , Privación de Alimentos , Humanos , Hipotálamo/metabolismo , Neuronas/metabolismo
17.
Physiol Behav ; 224: 113039, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32610101

RESUMEN

Dysfunction in neurophysiological systems that regulate food intake and metabolism are at least partly responsible for obesity and related comorbidities. An important component of this process is the hypothalamic melanocortin system, where an imbalance can result in severe obesity and deficits in glucose metabolism. Exercise offers many health benefits related to cardiovascular improvements, hunger control, and blood glucose homeostasis. However, the molecular mechanism underlying the exercise-induced improvements to the melanocortin system remain undefined. Here, we review the role of the melanocortin system to sense hormonal, nutrient, and neuronal signals of energy status. This information is then relayed onto secondary neurons in order to regulate physiological parameters, which promote proper energy and glucose balance. We also provide an overview on the effects of physical exercise to induce biophysical changes in the melanocortin circuit which may regulate food intake, glucose metabolism and improve overall metabolic health.


Asunto(s)
Metabolismo Energético , Melanocortinas , Homeostasis , Humanos , Hipotálamo , Obesidad
18.
Neuron ; 103(5): 891-908.e6, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31277924

RESUMEN

Motivated behavior is influenced by neural networks that integrate physiological needs. Here, we describe coordinated regulation of hypothalamic feeding and midbrain reward circuits in awake behaving mice. We find that alcohol and other non-nutritive drugs inhibit activity in hypothalamic feeding neurons. Interestingly, nutrients and drugs utilize different pathways for the inhibition of hypothalamic neuron activity, as alcohol signals hypothalamic neurons in a vagal-independent manner, while fat and satiation signals require the vagus nerve. Concomitantly, nutrients, alcohol, and drugs also increase midbrain dopamine signaling. We provide evidence that these changes are interdependent, as modulation of either hypothalamic neurons or midbrain dopamine signaling influences reward-evoked activity changes in the other population. Taken together, our results demonstrate that (1) food and drugs can engage at least two peripheral→central pathways to influence hypothalamic neuron activity, and (2) hypothalamic and dopamine circuits interact in response to rewards.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Inhibidores de Captación de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Etanol/farmacología , Conducta Alimentaria/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Agonistas Nicotínicos/farmacología , Recompensa , Proteína Relacionada con Agouti/metabolismo , Anfetamina/farmacología , Animales , Cocaína/farmacología , Antagonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/metabolismo , Hipotálamo/metabolismo , Ratones , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nicotina/farmacología , Proopiomelanocortina/metabolismo , Vagotomía , Nervio Vago/fisiología
19.
Aging Cell ; 18(5): e13014, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31373126

RESUMEN

The mechanistic target of rapamycin (mTOR) is an evolutionarily conserved protein kinase that regulates growth and metabolism. mTOR is found in two protein complexes, mTORC1 and mTORC2, that have distinct components and substrates and are both inhibited by rapamycin, a macrolide drug that robustly extends lifespan in multiple species including worms and mice. Although the beneficial effect of rapamycin on longevity is generally attributed to reduced mTORC1 signaling, disruption of mTORC2 signaling can also influence the longevity of worms, either positively or negatively depending on the temperature and food source. Here, we show that loss of hypothalamic mTORC2 signaling in mice decreases activity level, increases the set point for adiposity, and renders the animals susceptible to diet-induced obesity. Hypothalamic mTORC2 signaling normally increases with age, and mice lacking this pathway display higher fat mass and impaired glucose homeostasis throughout life, become more frail with age, and have decreased overall survival. We conclude that hypothalamic mTORC2 is essential for the normal metabolic health, fitness, and lifespan of mice. Our results have implications for the use of mTORC2-inhibiting pharmaceuticals in the treatment of brain cancer and diseases of aging.


Asunto(s)
Hipotálamo/metabolismo , Longevidad , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
20.
Neuron ; 100(6): 1277-1279, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30571939

RESUMEN

The brain orchestrates a variety of responses to noxious environmental stimuli, from reflexive movements to coordinated defensive behaviors. In this issue of Neuron, Barik et al. identify a hindbrain circuit essential for escape behaviors (Barik et al., 2018).


Asunto(s)
Calor , Rombencéfalo , Animales , Conducta Animal , Tronco Encefálico , Reacción de Fuga
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA